Elsevier

Brain Research

Volume 825, Issues 1–2, 17 April 1999, Pages 86-94
Brain Research

Research report
The febrile response to lipopolysaccharide is blocked in cyclooxygenase-2−/−, but not in cyclooxygenase-1−/− mice

https://doi.org/10.1016/S0006-8993(99)01225-1Get rights and content

Abstract

Various lines of evidence have implicated inducible cyclooxygenase-2 (COX-2) in fever production. Thus, its expression is selectively enhanced in brain after peripheral exogenous (e.g., lipopolysaccharide [LPS]) or endogenous (e.g., interleukin-1) pyrogen administration, while selective COX-2 inhibitors suppress the fever induced by these pyrogens. In this study, we assessed the febrile response to LPS of congenitally constitutive COX-1 (COX-1−/−) and COX-2 (COX-2−/−)-deficient C57BL/6J-derived mice. COX-1+/− and COX-2+/− mice were also evaluated; controls were wild-type C57BL/6J mice (Jackson Labs.). All the animals were pretrained daily for two weeks to the experimental procedures. LPS was injected intraperitoneally at 1 μg/mouse; pyrogen-free saline (PFS) was the vehicle and control solution. Core temperatures (Tcs) were recorded using thermocouples inserted 2 cm into the colon. The presence of the COX isoforms was determined in cerebral blood vessels immunocytochemically after the experiments, without knowledge of the functional results. The data showed that the wild-type, COX-1+/−, and COX-1−/− mice all responded to LPS with a 1°C rise in Tc within 1 h; the fever gradually abated over the next 4 h. By contrast, COX-2+/− and COX-2−/− mice displayed no Tc rise after LPS. PFS did not affect the Tc of any animal. It would appear therefore that COX-2 is necessary for LPS-induced fever production.

Introduction

Much evidence has accumulated indicating that prostaglandin E2 (PGE2) is the proximal mediator of fever (reviewed in Refs. 1, 13). It is believed to be produced and released in consequence of an action of pyrogenic cytokines (endogenous pyrogens, EnP) which are themselves produced and released in response to invading infectious pathogens or to their products, exogenous pyrogens (ExP). PGE2 is implicated as a fever mediator because, briefly: (1) it is a potent hyperthermic agent 1, 13thought to act directly or indirectly on thermoregulatory neurons in the preoptic anterior hypothalamus (POA), the primary brain site in which body temperature is regulated [2]; (2) its level increases and decreases in this brain region in conjunction with the febrile course 14, 43; (3) COX inhibitors, e.g., indomethacin and related nonsteroidal anti-inflammatory drugs (NSAIDs), inhibit pyrogen fever, in parallel with the reversal of PGE2 synthesis [30]; and (4) the congenital absence of the PGE2 EP3 receptor impairs the febrile response to both ExP and EnP [47]. Although PGE2 rises in blood promptly after the entry of microorganisms or after systemic ExP or EnP administration 31, 39, it is now generally agreed that the PGE2 detected in the brain is not derived from the blood, but rather is produced directly in the brain 15, 33, 41, albeit that some species differences may exist [17]. However, the cell source and nature of the triggering mechanism that induces PGE2 in the brain in response to systemic pyrogens, as well as its precise mode of action, remain controversial.

PGE2 is formed by the cleavage of membrane phospholipids by phospholipases, yielding arachidonic acid (AA). The released AA, in turn, is converted into the prostaglandin endoperoxides, PGG2 and PGH2, via sequential cyclisation and oxygenation by cyclooxygenase (COX) and peroxidation by hydroperoxidase; these two enzymatic activities co-exist in a single protein, prostaglandin H synthase [44]. PGH2 is then quickly isomerized to PGE2 by PGE2 isomerase. It is now recognized that there are at least two isoforms of COX which may [34]or may not [45]differ in their tissue and intracellular distributions, but are activated by distinct mechanisms. Thus, COX exists as a constitutive, COX-1, and an inducible, COX-2, isoenzyme; each is encoded by separate genes [35], but the enzymes share 60 to 70% homology 24, 50. COX-2 is up-regulated by proinflammatory mediators in, among other cell types, stimulated, but not unstimulated, macrophages and endothelial cells; it is selectively down-regulated by anti-inflammatory glucocorticoids. COX-2 is, however, also expressed constitutively in unstimulated neurons 4, 5, 6, 20, 25, 36, 37, 49, but the data differ on whether it is also up-regulated by pyrogenic stimuli 3, 11, 16, 18. By contrast, COX-1 is constitutively expressed in most cells; it is not affected by inflammatory mediators, and its basal activity is not altered by glucocorticoids (reviewed in Refs. 22, 23, 46).

Because of its inducibility by inflammatory mediators, it may be anticipated that COX-2 should have a major role in the brain in fever production. Indeed, it is now well-documented that ExP (e.g., bacterial endotoxic lipopolysaccharides [LPS]) and EnP (e.g., interleukin-1β [IL-1β]) activate COX-2 in vivo. Thus, in recent studies, COX-2-like immunoreactivity [28]and COX-2 mRNA 10, 11, 18, 37were found to be expressed in rat cerebral microvascular endothelial cells, particularly venules, ca. 1.5 h after intraperitoneal (i.p.) LPS and in perivascular microglia and meningeal macrophages ca. 2.5 h after intravenous (i.v.) LPS administration. In contrast, COX-1 expression was not affected anywhere in the brain by the peripheral administration of pyrogens. Moreover, treatment with specific inhibitors of COX-2 (NS-398, L-745, 337, DFU) orally after i.v. LPS [21]or intraperitoneally before i.p. LPS 11, 12, 38suppressed the febrile response, but did not affect basal body temperature. These antipyretic effects were not different from those produced by conventional NSAIDs, which inhibit both COX-1 and COX-2. Taken together, therefore, these data would seem to provide compelling support for the critical importance of COX-2 in fever genesis. To substantiate this inference, we examined the febrile response to LPS administered intraperitoneally of COX-1 and COX-2 gene knockout mice.

Section snippets

Animals

COX-1 and -2 gene heterozygous (COX-1+/−, COX-2+/−) and homozygous (COX-1−/−, COX-2−/−) knockout C57BL/6J-derived 27, 32and wild-type C57BL/6J mice (all 25–35 g in body weight) were used in these experiments. Following their transfer from the breeding area, the animals were quarantined for two weeks, four to a cage, before any experimental use. Tap water and food (Agway Prolab® mouse diet) were available ad libitum. The ambient temperature (Ta) in the animal room was 22±1°C; light and darkness

The thermal response

The i.p. injection procedure and associated handling rapidly induced a transient, ca. 1°C rise in the Tc of the wild-type mice, despite their training. It abated, however, over the following 45 min in the PFS-treated group (Fig. 1, open symbols). LPS administration, on the other hand, prevented this recovery, as evidenced by the sustained 1°C Tc rise over the first ca. 1.5 h (Fig. 1, closed symbols). The fever gradually abated over the next 2.5 h. The responses to the PFS and LPS treatments

Discussion

The present results are the first to demonstrate that COX-2 gene-deficient mice are unable to develop a full fever in response to the i.p. administration of a pyrogenic dose of LPS. The extent of the diminution of the febrile response appears to be proportional to the reduction in COX-2 expressed by these animals, as visualized by immunostaining of their cerebrovascular venular endothelial cells. The trend toward a more moderate, LPS-induced fall in Tc of the COX-2+/− than of the COX-2−/− mice

Acknowledgements

This study was supported, in part, by NIH grant NS 34857 to Clark M. Blatteis. We thank Dr. Y. Watanabe (Osaka Bioscience Institute) for his valuable comments and Mr. M. Ozaki for his technical assistance in the immunohistochemical analyses.

References (50)

  • A. Morimoto et al.

    Does an increase in prostaglandin E2 in the blood circulation contribute to a febrile response in rabbits?

    Brain Res. Bull.

    (1992)
  • I. Morita et al.

    Different intracellular locations for prostaglandin endoperoxide H synthase-1 and -2

    J. Biol. Chem.

    (1995)
  • N. Quan et al.

    Cyclooxygenase-2 mRNA expression in rat brain after peripheral injection of lipopolysaccharide

    Brain Res.

    (1998)
  • D. Rotondo et al.

    Pyrogenic immunomodulators increase the level of prostaglandin E2 in the blood simultaneously with the onset of fever

    Eur. J. Pharmacol.

    (1988)
  • E. Sehic et al.

    Blockade of lipopolysaccharide-induced fever by subdiaphragmatic vagotomy in guinea pigs

    Brain Res.

    (1996)
  • E. Sehic et al.

    Hypothalamic prostaglandin E2 during lipopolysaccharide-induced fever in guinea pigs

    Brain Res. Bull.

    (1996)
  • W.L. Smith et al.

    Prostaglandin endoperoxide H synthase-1 and -2

    Adv. Immunol.

    (1996)
  • K. Yamagata et al.

    Expression of a mitogen-inducible cyclooxygenase in brain neurons: regulation by synaptic activity and glucocorticoids

    Neuron

    (1993)
  • C. Blatteis, E. Sehic, Prostaglandin E2: a putative fever mediator, in: P.A. Mackowiak (Ed.), Fever: Basic Mechanisms...
  • J.A. Boulant, Hypothalamic neurons regulating body temperature, in: M.J. Fregly, C.M. Blatteis (Eds.), Handbook of...
  • C.D. Breder

    Cyclooxygenase systems in the mammalian brain

    Ann. New York Acad. Sci.

    (1997)
  • C.D. Breder et al.

    Characterization of inducible cyclooxygenase in rat brain

    J. Comp. Neurol.

    (1995)
  • C.D. Breder et al.

    Distribution and characterization of cyclooxygenase immunoreactivity in the ovine brain

    J. Comp. Neurol.

    (1992)
  • A. Cabanac et al.

    Handling elevates the colonic temperature of mice

    Physiol. Behav.

    (1991)
  • C. Cao et al.

    Involvement of cyclooxygenase-2 in LPS-induced fever and regulation of its mRNA by LPS in the rat brain

    Am. J. Physiol.

    (1997)
  • Cited by (232)

    • Reflex regulation of systemic inflammation by the autonomic nervous system

      2022, Autonomic Neuroscience: Basic and Clinical
      Citation Excerpt :

      There is evidence that this can happen: the relevant receptors are present on CVO neurons, and both functional studies and Fos immunohistochemistry indicate excitatory responses in those neurons (Korim et al., 2019; Roth et al., 2004; Wei et al., 2018; Wei et al., 2013). As lipids, prostaglandins are not excluded by the blood brain barrier and may also signal directly to the brain, as indeed they do to trigger fever (Engstrom et al., 2012; Lazarus et al., 2007; Li et al., 1999; Saper, 1998). On the other hand, it is understood that peripheral nerves can carry relevant inflammatory signals to the CNS (Goehler et al., 2000; Niijima, 1996; Ross et al., 2000; Soto-Tinoco et al., 2020).

    View all citing articles on Scopus
    View full text